WO2006094068A2 - Inhibiteurs d’hdac favorisant l’expression des brm et diagnostics associes aux brm - Google Patents

Inhibiteurs d’hdac favorisant l’expression des brm et diagnostics associes aux brm Download PDF

Info

Publication number
WO2006094068A2
WO2006094068A2 PCT/US2006/007296 US2006007296W WO2006094068A2 WO 2006094068 A2 WO2006094068 A2 WO 2006094068A2 US 2006007296 W US2006007296 W US 2006007296W WO 2006094068 A2 WO2006094068 A2 WO 2006094068A2
Authority
WO
WIPO (PCT)
Prior art keywords
brm
expression
protein
assay
cell
Prior art date
Application number
PCT/US2006/007296
Other languages
English (en)
Other versions
WO2006094068A3 (fr
Inventor
David Reisman
Original Assignee
The Regents Of The University Of Michigan
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Regents Of The University Of Michigan filed Critical The Regents Of The University Of Michigan
Publication of WO2006094068A2 publication Critical patent/WO2006094068A2/fr
Publication of WO2006094068A3 publication Critical patent/WO2006094068A3/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57496Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving intracellular compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/24Immunology or allergic disorders

Definitions

  • the present invention relates to methods and compounds for causing BRM re- expression in cells, such as cancer cells, that have lost BRM expression.
  • the present invention relates to screening methods for identifying BRM expression-promoting compounds, such as histone deacetylase inhibitors (HDAC) inhibitors; diagnostic methods for determining the suitability of treatment of a candidate subj ect with a BRM expression- promoting HDAC inhibitor; and therapeutic methods for treating cancer cells in a patient with a BRM expression-promoting HDAC inhibitor.
  • HDAC histone deacetylase inhibitors
  • the present invention also relates to BRGl and BRM diagnostics, methods for increasing a cancer patient's resistance to viral infection, methods for monitoring therapy, and methods for determining the suitability of treatment of a candidate subject with a glucocorticoid compound or retinoid compound.
  • the present invention provides screening methods for identifying BRM expression- promoting histone deacetylase (HDAC) inhibitors, diagnostic methods for determining the suitability of treatment of a candidate subject with a BRM expression-promoting HDAC inhibitor, and therapeutic methods for treating cancer cells in a patient with a BRM expression-promoting HDAC inhibitor.
  • the present invention also provides BRGl and BRM diagnostics, methods for monitoring therapy, methods for increasing a cancer patient's resistance to viral infection, and methods for determining the suitability of treatment of a candidate subject with a glucocorticoid compound or retinoid compound.
  • the present invention provides methods of identifying a BRM expression-promoting histone deacetylase inhibitor comprising; a) providing; i) a candidate histone deacetylase inhibitor; and ii) at lease one cell (e.g., a plurality of cells), wherein the cell exhibits reduced BRM protein or BRM mRNA expression; b) contacting the cell with the candidate histone deacetylase inhibitor, and c) measuring BRM protein or BRM mRNA expression exhibited by the cell, or measuring BRM-regulated protein or BRM-regulated mRNA expression from a BRM regulated gene exhibited by the cell, wherein an increase in the BRM protein, BRM mRNA expression, BRM-regulated protein expression, or BRM- regulated mRNA expression exhibited by the cell identifies the candidate histone deacetylase inhibitor as a BRM expression-promoting histone deactylase inhibitor.
  • the BRM regulated gene is a gene shown in Table 4.
  • the BRM expression-promoting histone deacetylase inhibitor inhibits a human histone deacetylase protein selected from the group consisting of: HDACl, HDAC2, HDAC3, HDAC8, and HDACIl. In other embodiments, the BRM expression-promoting histone deacetylase inhibitor inhibits a human histone deacetylase protein selected from the group consisting of: HDAC4, HDAC5, HDAC7, and HDAC9. In other embodiments, the BRM expression-promoting histone deacetylase inhibitor inhibits HDACl.
  • the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDACl. In some embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDAC2. In other embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDAC3. In additional embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDAC4. hi further embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human BDD AC5. In particular embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDAC6. In other embodiments, the BRM expression- promoting histone deacetylase inhibitor specifically inhibits human HDAC7.
  • the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDAC8. In particular embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDAC9. In other embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDAClO. hi some embodiments, the BRM expression-promoting histone deacetylase inhibitor specifically inhibits human HDACl 1.
  • the candidate histone deacetylase inhibitor is identified as a BRM expression-promoting histone deactylase inhibitor, and the method further comprises step d) determining if the BRM protein expressed by the cell after the contacting is active or inactive BRM protein, wherein only the active BRM protein can form a functioning SWI/SNF complex in the cell, hi some embodiments, determining if the BRM protein expressed by the cells is active or inactive BRM protein comprises performing an assay to determine if PPARgamma, CD44 or vimentin is up-regulated in the cell, hi additional embodiments, the method further comprises step d) determining if CD44 or vimentin is up-regulated in the cell, hi other embodiments, the method further comprises step d) measuring retinoblastoma protein growth inhibition in the cell, hi some embodiments, the methods further comprises step d) determining if p53, plO7, BRCAl or Farconi's anemia protein are expressed by the cell, hi particular embodiments, the B
  • the cell further exhibits reduced wild-type BRGl protein or wild-type BRGl mRNA expression.
  • the candidate histone deacetylase inhibitor is selected from the group consisting of: a short chain fatty acid, a hydroxamic acid, a tetrapeptide, and a cyclic hydroxamic acid containing peptide, hi preferred embodiments, the candidate histone deacetylase inhibitor is selected from the group consisting of: apicidin, butyrates, depsipeptide, FR901228, FK-228, Depudecin, m- carboxy cinnamic acid, bishydroxamic acid, MS-275, N-acetyl dinaline, oxamflatin, pyroxamide, sciptaid, suberoylanilie hydroxamic acid, TPX-HA analogue (CHAP), trapoxin, trichostatin A, and, SB-79872, SB-29201, tabuci
  • the cell is a cancer cell.
  • the cancer cell is breast cancer cell or a prostate cancer cell (e.g. a hormone insensitive prostate cancer cell).
  • the cell is from a cell line selected from the group consisting of: H513, H522, H23, H125, A427, SW13, C33A, Panc-1, H1573, and H1299.
  • the cell exhibits reduced BRM protein expression
  • the cell exhibits reduced BRM niRNA expression.
  • the cell is a human cell.
  • the cell is part of an animal model (e.g. the cell is part of a tumor growing on or in an animal, such as a mouse or rat).
  • contacting the cell with the candidate histone deacetylase inhibitor is performed in a microtiter plate (e.g. a 96-well plate). In some embodiments, contacting the cell with the candidate histone deacetylase inhibitor is performed in an automated fashion (e.g. for high-throughput screening).
  • the measuring BRM protein or BRM mRNA expression comprises measuring the BRM protein expression
  • the BRM protein expression comprises performing an ELISA assay, a Western Blot, or any other type of protein detection assay.
  • the protein detection assay employs an anti-BRM antibody.
  • the measuring BRM protein or BRM mRNA expression comprises measuring the BRM mRNA expression.
  • measuring the mRNA expression comprises a detection assay selected from the group consisting of: an INVADER assay, a TAQMAN assay, a sequencing assay, a polymerase chain reaction assay, a hybridization assay, a hybridization assay employing a probe complementary to a mutation, a microarray assay, a bead array assay, a primer extension assay, an enzyme mismatch cleavage assay, a branched hybridization assay, a rolling circle replication assay, a NASBA assay, a molecular beacon assay, a cycling probe assay, a ligase chain reaction assay, and a sandwich hybridization assay.
  • the present invention provides methods for identifying a BRM expression-promoting compound comprising; a) providing; i) a candidate compound; and ii) at least one cell (e.g., plurality of cells), wherein the cell exhibits reduced BRM protein or BRM mRNA expression; b) contacting the cell with the candidate compound, and c) measuring BRM protein or BRM mRNA expression exhibited by the cell, or measuring BRM-regulated protein or BRM-regulated mRNA expression from a BRM regulated gene exhibited by the cell, wherein an increase in the BRM protein, BRM mRNA expression, BRM-regulated protein expression, or BRM-regulated mRNA expression, exhibited by the cell identifies the candidate compound as a BRM expression-promoting compound.
  • the BRM regulated gene is a gene shown in Table 4.
  • the candidate compound is identified as a BRM expression- promoting compound, and the method further comprises step d) determining if the BRM protein expressed by the cell after the contacting is active or inactive BRM protein, wherein only the active BRM protein can form a functioning SWI/SNF complex in the cell.
  • the BRM protein is determined to be the active BRM protein thereby indicating that the BRM expression-promoting compound is an active BRM expression- promoting compound.
  • the present invention provides methods of determining the suitability of treatment of a candidate subject with a BRM expression-promoting histone deacetylase inhibitor, comprising; a) providing a plurality of cancer cells from a candidate subject; b) measuring BRM protein or BRM mRNA expression exhibited by the plurality of cancer cells, or measuring BRM-regulated protein or BRM-regulated mRNA expression from a BRM regulated gene, exhibited by the plurality of cancer cells, in order to determine if the plurality of cancer cells exhibit wild-type or reduced expression of the BRM protein; and c) determining the suitability of treating the candidate subject with a BRM expression- promoting histone deacetylaste inhibitor, wherein the candidate subject is suitable for such treatment if it is determined that the plurality of cells exhibit reduced expression of the BRM protein or the BRM mRNA.
  • the BRM regulated gene is a gene shown in Table 4.
  • the present invention provides methods of identifying a candidate subject as suitable for treatment with a BRM expression-promoting histone deactylase inhibitor, comprising; a) providing a plurality of cancer cells from a candidate subject; b) measuring BRM protein or BRM mRNA expression exhibited by the plurality of cancer cells, or measuring BRM-regulated protein or BRM-regulated mRNA expression from a BRM regulated gene, exhibited by the plurality of cancer cells, in order to determine if the plurality of cancer cells exhibit wild-type or reduced expression of the BRM protein, and c) identifying the candidate subject as suitable for treatment with a BRM expression- promoting histone deacetylase inhibitor, wherein the identifying comprises finding that the plurality of cells exhibit reduced expression of the BRM protein or the BRM mRNA.
  • the BRM regulated gene is a gene shown in Table 4.
  • the plurality of cells further exhibit reduced wild-type
  • the methods further comprise a step of determining if CD44 or vimentin is up-regulated in the cell.
  • the present invention provides methods of identifying a candidate subject suitable for treatment with a BRM expression-promoting compound, comprising; a) providing a plurality of cancer cells from a candidate subject; b) measuring BRM protein or BRM mRNA expression exhibited by the plurality of cancer cells, and c) identifying the candidate subject as suitable for treatment with a BRM expression- promoting compound, wherein the identifying comprises finding that the plurality of cells exhibit reduced expression of the BRM protein or the BRM mRNA.
  • the plurality of cancer cells comprise a biopsy sample from the candidate subject.
  • the present invention provides methods of treating cancer cells in a patient comprising; a) identifying a patient comprising a plurality cancer cells, wherein the plurality of cancer cells exhibit reduced BRM protein or BRM mRNA expression; and b) administering a BRM expression-promoting histone deacetylate inhibitor to the patient under conditions such that at least a portion of the plurality of cancer cells are killed, hi certain embodiments, the methods further comprise c) administering a glucocorticoid compound or a retinoid compound to the patient, hi some embodiments, the glucocorticoid compound is selected from the group consisting of: hydrocortisone, prenisone (deltasone), predrisonlone (hydeltasol), Cortisol (hydrocortisone), dexamethasone, triamcinolone, betamethasone, beclomethasone, methylprednisolone, fludrocortisone acetate, deoxycorticosterone
  • the present invention provides methods of treating cancer cells in a patient comprising; a) identifying a patient comprising a plurality cancer cells, wherein the plurality of cancer cells are suspected of having reduced BRM protein or BRM mRNA expression; and b) administering a BRM expression-promoting histone deacetylate inhibitor to the patient under conditions such that at least a portion of the plurality of cancer cells are killed.
  • the methods further comprise c) administering a glucocorticoid compound or a retinoid compound to the patient.
  • the glucocorticoid compound is selected from the group consisting of: hydrocortisone, prenisone (deltasone), predrisonlone (hydeltasol), Cortisol (hydrocortisone), dexamethasone, triamcinolone, betamethasone, beclomethasone, methylprednisolone, fludrocortisone acetate, deoxycorticosterone acetate (DOCA), and aldosterone.
  • hydrocortisone prenisone
  • deltasone predrisonlone
  • hydeltasol predrisonlone
  • Cortisol hydrocortisone
  • dexamethasone triamcinolone
  • betamethasone beclomethasone
  • methylprednisolone fludrocortisone acetate
  • deoxycorticosterone acetate DHA
  • aldosterone aldosterone
  • the retinoid compound is selected from the group consisting of: retinoid - 9-cis retinoic acid, vitamin A, retinaldehyde, retinol, retinoic acid, tretinoin,, iso-tretinoin, and related compounds.
  • the present invention provides methods of treating cancer cells in a patient comprising; a) identifying a patient comprising a plurality cancer cells, wherein the plurality of cancer cells exhibit reduced BRM protein or BRM mRNA expression; and b) administering a BRM expression-promoting histone deacetylate inhibitor to the patient under conditions such that a least a portion of the plurality of cancer cells express active BRM protein thereby allowing functional SWI/SNF complexes to form in the plurality of cells.
  • the BRM expression-promoting histone deacetylase inhibitor is an active BRM expression-promoting histone deacetylase inhibitor.
  • the methods further comprise c) administering a glucocorticoid compound or a retinoid compound to the patient.
  • the present invention provides methods of treating cancer cells in a patient comprising; a) providing; i) a composition comprising; A) a plurality of BRM proteins, or B) an expression vector configured to express a BRM protein; and ii) a patient comprising a plurality cancer cells suspected of, or having, reduced BRM protein expression; and b) administering the composition to the patient under conditions such that at least a portion of the plurality of cancer cells are killed.
  • the expression vector comprises a nucleic acid sequence encoding the BRM protein.
  • the methods further comprise c) administering a glucocorticoid compound or a retinoid compound to the patient.
  • the present invention provides methods of treating cancer cells in a patient comprising; a) providing; i) a composition comprising a nucleic acid sequence configured to interfere with expression of a histone deacetylase, and ii) a patient comprising a plurality of cancer cells suspected of, or having, reduced BRM protein expression; and b) administering the composition to the patient under conditions such that at least a portion of the plurality of cancer cells are killed.
  • the nucleic acid sequence comprises siRNA or antisense directed against the histone deacetylase.
  • the present invention provides methods for determining the suitability of treatment of a candidate subject with a glucocorticoid compound or retinoid compound, comprising; a) providing a plurality of cancer cells from a candidate subject; b) measuring BRM protein or BRM mRNA expression exhibited by the plurality of cancer cells in order to determine if the plurality of cancer cells exhibit wild-type or reduced expression of the BRM protein; and c) determining the suitability of treating the candidate subject with a glucocorticoid compound or retinoid compound, wherein the candidate subj ect is suitable for such treatment if it is determined that the plurality of cells exhibit wild-type expression of the BRM protein.
  • the present invention provides methods of determining the suitability of treatment of a candidate subject with a glucocorticoid compound or retinoid compound, comprising; a) providing a plurality of cancer cells from a candidate subject; b) measuring BRM protein expression, BRM mRNA expression, or measuring BRM-regulated protein or BRM-regulated mRNA expression of a BRM regulated gene, exhibited by the plurality of cancer cells in order to determine if the plurality of cancer cells exhibit wild-type or reduced expression of the BRM protein; and c) determining the suitability of treating the candidate subject with a glucocorticoid compound or retinoid compound, wherein the candidate subject is suitable for such treatment if it is determined that the plurality of cells exhibit wild-type expression of the BRM protein.
  • the BRM regulated gene is a gene shown in Table 4.
  • the plurality of cells are determined to exhibit wild-type expression of the BRM protein, and wherein the method further comprises d) administering the glucocorticoid compound or the retinoid compound to the candidate subject. In further embodiments, the plurality of cells are determined to exhibit reduced expression of the BRM protein, and wherein the method further comprises d) administering both a histone deacetylase inhibitor and the glucocorticoid compound or the retinoid compound to the candidate subject. In other embodiments, the plurality of cells are determined to exhibit reduced expression of the BRM protein, and the patient'is identified as not suitable for treatment by the glucocorticoid compound or the retinoid compound (e.g.
  • the patient's records are marked as not suitable for treatment with glucocoriticoid or retinoid compounds).
  • the glucocorticoid compound is selected from the group consisting of: hydrocortisone, prenisone (deltasone), predrisonlone (hydeltasol), Cortisol (hydrocortisone), dexamethasone, triamcinolone, betamethasone, beclomethasone, methylprednisolone, fludrocortisone acetate, deoxycorticosterone acetate (DOCA), and aldosterone.
  • hydrocortisone prenisone
  • deltasone predrisonlone
  • hydeltasol predrisonlone
  • Cortisol hydrocortisone
  • dexamethasone triamcinolone
  • betamethasone beclomethasone
  • methylprednisolone fludrocortisone acetate
  • the retinoid compound is selected from the group consisting of: retinoid - 9-cis retinoic acid, vitamin A, retinaldehyde, retinol, retinoic acid, tretinoin,, iso-tretinoin, and related compounds.
  • the retinoid compound comprises Bexarotene (e.g. TARGRETIN).
  • the present invention provides methods of increasing a cancer patient's resistance to viral infection, wherein the cancer patient comprises a plurality of cancer cells, the method comprising administering a BRM expression-promoting histone deacetylase inhibitor to the cancer patient under conditions such that expression of at least one interferon-induced gene (e.g. as shown in Table 4) is up-regulated in the plurality of cancer cells thereby increasing the cancer patient's resistance to viral infection.
  • the interferon-induced gene is up-regulated as least 4-fold.
  • the BRM expression-promoting histone deacetylase inhibitor is co- administered with a cancer therapy, such as chemotherapy, radiation, surgery, etc.
  • the cancer patient is undergoing treatment with one or more therapeutic compounds that reduce the cancer patient's resistance to viral infection.
  • the therapeutic compounds is a glucocorticoid compound or a retinoid compound.
  • the present invention provides methods of increasing a cancer patient's resistance to viral infection, wherein the cancer patient comprises a plurality of cancer cells, the method comprising administering i) a plurality of BRM proteins, or ii) an expression vector configured to express a BRM protein, to the cancer patient under conditions such that expression of at least one interferon-induced gene is up-regulated in the plurality of cancer cells thereby increasing the cancer patient's resistance to viral infection, hi some embodiments, the cancer patient is undergoing treatment with one or more therapeutic compounds that reduce the cancer patient's resistance to viral infection.
  • the therapeutic compounds is a glucocorticoid compound or a retinoid compound.
  • the present invention provides methods comprising: a) providing a sample comprising a nucleic acid sequence, wherein the nucleic acid sequence comprises at least a portion of a BRM gene or a BRGl gene; and b) contacting the sample with a nucleic acid detection assay under conditions such that the presence or absence of a SWI/SNF complex formation polymorphism (e.g. a polymorphism that, if present, prevents the successful formation of the SWI/SNF complex) is detected in the BRM gene or the BRGl gene.
  • a SWI/SNF complex formation polymorphism e.g. a polymorphism that, if present, prevents the successful formation of the SWI/SNF complex
  • the nucleic acid sequence comprises an amplification product.
  • the amplification product comprises a PCR amplification product.
  • the nucleic acid detection assay is selected from the group consisting of: a TAQMAN assay, an invasive cleavage assay, a sequencing assay, a polymerase chain reaction assay, a hybridization assay, a microarray assay, a bead array assay, a primer extension assay, an enzyme mismatch cleavage assay, a branched hybridization assay, a rolling circle replication assay, a NASBA assay, a molecular beacon assay, a cycling probe assay, a ligase chain reaction assay, a sandwich hybridization assay, and a Line Probe Assay.
  • the nucleic acid sequence is derived from a cancer cell.
  • the cancer cell is from a cancer patient (e.g. from a biopsy of a tumor from a
  • the nucleic acid sequence comprises a BRM promoter sequence, and the polymorphism is located at position 741 (as shown in Figure 5).
  • the polymorphism at position 741 is a 7 base pair insertion (e.g. TATTTTT; SEQ ID NO:42).
  • the nucleic acid sequence comprises at least a portion of the BRGl gene, and wherein the polymorphism causes an amino acid substitution selected from the group consisting of: P311S; P316S; P319S, and P327S (as shown in Figure IB).
  • the nucleic acid sequence is derived from a cancer cell, wherein the nucleic acid sequence comprises a BRM promoter sequence, and the polymorphism is located at position 741.
  • the cell is determined to be heterozygous or homozygous for the position 741 polymorphism.
  • the present invention provides compositions comprising an isolated nucleic sequence that comprises SEQ ID NO: 52
  • the nucleic acid sequence serves as a positive control for a nucleic acid detection assay configured to detect the presence of the seven base pair insertion in the BRM promoter shown in Figure 5.
  • Figure 1 shows the location of various BRGl mutations.
  • Figure IA illustrates the location of each alteration detected in the BRGl gene with respect to the known domains. Unshaded triangles below the domains represent splicing defects. The circles denote sites of deletions and the hexagons denote the sites of nonsense mutations.
  • Figure IB shows missense mutations in a proline-rich region of BRGl . The illustrated region shows a 20- amino- acid region (SEQ ID NO:41) in the N-terminus of the BRGl gene, which is highly conserved across the human BRGl and BRM genes, as well as the BRGl genes of Xenopus, Drosophila, and Danio. In the cell lines C33A, Panc-1, H 1299, and SWl 3, the conserved prolines in this region are mutated to serines (denoted by arrows).
  • Figure 2 shows BRGl splicing defects in BRGl/BRM-deficient cell lines.
  • Figure 2 A shows sequencing chromatographs corresponding to each alteration found in the BRGl gene. The 69 bp deletion in H 1299 is represented by an agarose gel illustrating the truncated PCR product compared to a normal control. Each of the sequence changes appears homozygous, as the unaltered wild-type allele was not detected.
  • Figure 2B shows the location of the BRGl splicing defect in the H513, H23, and H 1299 cell lines, which resulted in 718, 386, and 250 bp deletions in BRGl, as illustrated in the left column. The junction of each splicing variant is depicted in the chromatograph on the right. The different exons are shaded and labeled. Each aberrantly spliced variant alters the reading frame upstream of the ATPase domain.
  • Figure 3 shows the temporal effects of the small molecular inhibitor sodium butyrate on BRM expression.
  • Figure 3 A shows BRM protein re-expression in sodium butyrate- treated cell lines. Cells were treated daily with sodium butyrate (5mM). Total protein was extracted at 4, 12, 24, 36, 50, and 72 hours after the first dosage. Upregulation of BRM with butyrate treatment was detected after 12 hours and reached a plateau between 24 and 48 hours. GAPDH was the loading control.
  • Figure 4 A shows the experimental design of the mouse breeding and sequential treatment with the lung-specific carcinogen, urethane, described in Example 6.
  • Figure 4B shows that the number of tumors in the mice 12 weeks post urethane treatment for mice that were wild type, heterozygous or null for BRM expression. Compared to wild type mice, BRM heterozygous and BRM null had approximately 4- and 10-fold more tumors on the surface of the lung, respectively.
  • Figure 4C shows that when tumors were counted in cross sections, a 3- and 7-fold increase in tumors was found when one or both BRM alleles were missing.
  • Figure 5 show the nucleic acid sequence of the human BRM promoter with the seven base insert (SEQ ID NO:42) at position 741 underlined.
  • the terms “subject” and “patient” refer to any animal, such as a mammal like a dog, cat, bird, livestock, and preferably a human. Specific examples of “subjects” and “patients” include, but are not limited to, individuals with cancer, such as breast cancer or prostate cancer.
  • wild-type refers to a gene or protein that has the characteristics of that gene or gene product when isolated from a naturally occurring source.
  • a wild-type gene or protein is that which is most frequently observed in a population and is thus arbitrarily designed the "normal” or “wild-type” form of the gene.
  • variant refers to a gene or protein that displays modifications in sequence and/or functional properties (i.e., altered characteristics) when compared to the wild-type gene or gene product.
  • antisense is used in reference to RNA sequences that are complementary to a specific RNA sequence (e.g., mRNA).
  • Antisense RNA may be produced by any method, including synthesis by splicing the gene(s) of interest in a reverse orientation to a viral promoter that permits the synthesis of a coding strand. Once introduced into an embryo, this transcribed strand combines with natural mRNA produced by the embryo to form duplexes. These duplexes then block either the further transcription of the mRNA or its translation, hi this manner, mutant phenotypes may be generated.
  • the term “antisense strand” is used in reference to a nucleic acid strand that is complementary to the "sense” strand.
  • the designation (-) i.e., "negative" is sometimes used in reference to the antisense strand, with the designation (+) sometimes used in reference to the sense (i.e., "positive”) strand.
  • siRNAs refers to short interfering RNAs.
  • siRNAs comprise a duplex, or double-stranded region, of about 18-25 nucleotides long; often siRNAs contain from about two to four unpaired nucleotides at the 3' end of each strand.
  • At least one strand of the duplex or double-stranded region of a siRNA is substantially homologous to or substantially complementary to a target RNA molecule.
  • the strand complementary to a target RNA molecule is the "antisense strand;" the strand homologous to the target RNA molecule is the "sense strand,” and is also complementary to the siRNA antisense strand.
  • siRNAs may also contain additional sequences; non-limiting examples of such sequences include linking sequences, or loops, as well as stem and other folded structures. siRNAs appear to function as key intermediaries in triggering RNA interference in invertebrates and in vertebrates, and in triggering sequence-specific RNA degradation during posttranscriptional gene silencing in plants.
  • RNA interference refers to the silencing or decreasing of gene expression by siRNAs. It is the process of sequence-specific, post-transcriptional gene silencing in animals and plants, initiated by siRNA that is homologous in its duplex region to the sequence of the silenced gene.
  • the gene may be endogenous or exogenous to the organism, present integrated into a chromosome or present in a transfection vector that is not integrated into the genome. The expression of the gene is either completely or partially inhibited.
  • RNAi may also be considered to inhibit the function of a target RNA; the function of the target RNA may be complete or partial.
  • BRM regulated gene refers to any gene who's mRNA and/or protein expression is increased in a cell when BRM mRNA or protein expression is increased in said cell.
  • BRM regulated genes include, but are noted limited to, CD44, E-cadherin, SPARK, LBH, CEA CAM-I, S100A2, RARR3, GADD45a, an interferon induced gene, and genes shown in Table 4.
  • Southern blot refers to the analysis of DNA on agarose or acrylamide gels to fractionate the DNA according to size followed by transfer of the DNA from the gel to a solid support, such as nitrocellulose or a nylon membrane.
  • the immobilized DNA is then probed with a labeled probe to detect DNA species complementary to the probe used.
  • the DNA may be cleaved with restriction enzymes prior to electrophoresis. Following electrophoresis, the DNA may be partially depurinated and denatured prior to or during transfer to the solid support.
  • Southern blots are a standard tool of molecular biologists (J. Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, NY, pp 9.31-9.58 [1989]).
  • Northern blot refers to the analysis of RNA by electrophoresis of RNA on agarose gels to fractionate the RNA according to size followed by transfer of the RNA from the gel to a solid support, such as nitrocellulose or a nylon membrane. The immobilized RNA is then probed with a labeled probe to detect RNA species complementary to the probe used.
  • Northern blots are a standard tool of molecular biologists (J. Sambrook, et al, supra, pp 7.39-7.52 [1989]).
  • the term “Western blot” refers to the analysis of protein(s) (or polypeptides) immobilized onto a support such as nitrocellulose or a membrane.
  • the proteins are run on acrylamide gels to separate the proteins, followed by transfer of the protein from the gel to a solid support, such as nitrocellulose or a nylon membrane.
  • the immobilized proteins are then exposed to antibodies with reactivity against an antigen of interest.
  • the binding of the antibodies may be detected by various methods, including the use of radiolabeled antibodies.
  • the phrase “candidate compound” refer to any chemical entity, pharmaceutical, drug, and the like that can be used to treat or prevent a disease, illness, sickness, or disorder of bodily function, or otherwise alter the physiological or cellular status of a sample.
  • Test compounds comprise both known and potential therapeutic compounds.
  • a test compound can be determined to be therapeutic by screening using the screening methods of the present invention.
  • histone deacetylase and "HDAC” are intended to refer to any one of a family of enzymes that remove acetyl groups from the epsilon-amino groups of lysine residues at the N-terminus of a histone. Unless otherwise indicated by context, the term “histone” is meant to refer to any histone protein, including Hl, H2A, H2B, H3, H4, and H5, from any species. Preferred histone deacetylases include class I and class II enzymes.
  • the histone deacetylase is a human HDAC, including, but not limited to, HDAC-I, HDAC-2, HDAC-3, HDAC-4, HDAC-5, HDAC-6, HDAC-7, HDAC-8, HDAC-9, HDAC-10, and HDAC-11.
  • histone deacetylase inhibitor or "inhibitor of histone deacetylase” is used to identify a compound which is capable of interacting with a histone deacetylase and inhibiting its enzymatic activity. Inhibiting histone deacetylase enzymatic activity means reducing the ability of a histone deacetylase to remove an acetyl group from a histone. hi some preferred embodiments, such reduction of histone deacetylase activity is at least about 50%, more preferably at least about 75%, and still more preferably at least about 90%. In other preferred embodiments, histone deacetylase activity is reduced by at least 95% and more preferably by at least 99%.
  • such inhibition is specific, such that the histone deacetylase inhibitor reduces the ability of a histone deacetylase to remove an acetyl group from a histone at a concentration that is lower than the concentration of the inhibitor that is required to produce another, unrelated biological effect.
  • concentration of the inhibitor required for histone deacetylase inhibitory activity is at least 2-fold lower, more preferably at least 5-fold lower, even more preferably at least 10-fold lower, and most preferably at least 20-fold lower than the concentration required to produce an unrelated biological effect.
  • a "BRM expression-promoting histone deacetylase inhibitor” is a histone deacetylase inhibitor that is able to cause a cell with reduced BRM protein or niRNA expression to begin expressing BRM protein or mRNA, or increase the level of expression or BRM protein or mRNA (e.g. by at least 20%), when contacted with that cell.
  • a histone deacetylase inhibitor "specifically inhibits” a given HDAC when the inhibitor only inhibits the function of the given HDAC in a cell, and not any of the other HDACs. For example, if a histone deacetylase inhibitor "specifically inhibits" HDAC2 in a human cell, this inhibitor, when contacted with a cell, would not inhibit HDACs 1, 3, 4, 5, 6, 7, 8, 9, 10 and 11.
  • a cell exhibits "reduced BRM protein or BRM mRNA expression" when the cell either exhibits no BRM protein or mRNA expression, or the level of BRM protein or BRM mRNA expression is less than 75 percent of that wild type level found in cells of the same type (e.g. cells of the same type that are not cancerous).
  • a cell exhibits "reduced wild-type BRGl protein or wild-type BRGl mRNA expression" when the cells exhibits no wild-type BRGl protein or mRNA expression (e.g. all of the BRGl protein expressed is mutant form), or the level of wild-type BRGl protein or wild-type BRGl mRNA is less than 75 percent of the wild-type level found in cells of the same type (e.g. cells of the same type that are not cancerous).
  • suitable for treatment with a BRM expression-promoting histone deacetylase inhibitor when used in reference to a candidate subject refers to subjects who are more likely to benefit from such treatment than a subject selected randomly from the population.
  • An example of such a candidate subject is one who has been determined to have cancer cells with reduced BRM expression.
  • the present invention provides screening methods for identifying BRM expression- promoting histone deacetylase (HDAC) inhibitors, diagnostic methods for determining the suitability of treatment of a candidate subject with a BRM expression-promoting HDAC inhibitor, and thereapeutic methods for treating cancer cells in a patient with a BRM expression-promoting HDAC inhibitor.
  • the present invention also relates to BRGl and BRM diagnostics, methods for increasing a cancer patient's resistance to viral infection, and methods for determining the suitability of treatment of a candidate subject with a glucocorticoid compound or retinoid compound.
  • SWI/SNF Complex Chromatin remodeling plays an essential role in regulating gene expression. By controlling which areas of chromatin are open or condensed, cells are limited to which genes they can express. Along the chromatin, histones are marked by the addition of acetyl or methyl groups. These secondary modifications to histones provide a code (a histone code) that determines which specific areas of the chromatin will be opened or condensed. This histone code is maintained and read by a complex array of multimeric proteins collectively called chromatin remodeling complexes. Restricting the accessibility of the DNA in this way limits the function of transcription factors and key cellular proteins and is used by normal cells to maintain differentiation and control growth. However, cancer cells can escape these restraints by disrupting the function of these chromatin remodeling complexes.
  • the SWI/SNF complex is one such important chromatin remodeling complex that is involved in gene regulation and whose dysregulation has been shown to contribute to cancer development.
  • the SWI/SNF complex contains 9-12 proteins and provides direct access to DNA by shifting the position of the histones (Wang et al., Curr. Top. Microbiol. Immunol., 2003, 274: 143-69, herein incorporated by reference). It was first linked to tumorigenesis with the finding that the SWI/SNF subunit, BAF47, is a bonaflde tumor suppressor protein. The loss of this protein has been shown to be a key event in the development of rhabdoid sarcoma, a lethal pediatric tumor, hi cell lines derived from these tumors, re-expression of the BAF47 proteins causes pronounced growth arrest and differentiation.
  • This subunit is essential, as its loss disrupts function of the SWI/SNF complex.
  • BRM expression is restored in cancer cell lines, a progressive growth arrest ensues and the cells adopt a flattened, differentiated phenotype.
  • This observation supports the role of the SWI/SNF complex in facilitating growth- controlling pathways.
  • alterations to the SWI/SNF complex appear to occur in a number of tumor types. It has been found by immunostaining tissue microarrays (TMAs) that the expression of BRM is lost in 5-15 % of esophageal, ovarian, prostate, bladder, head/neck tumors and lung cancer. Which pathways are selectively disrupted when the SWI/SNF complex is abrogated is not currently known.
  • retinoic acid receptor (RAR) and proxisome proliferative receptor gamma (PP AR7) which oppose cancer development, require the SWI/SNF complex
  • tumor suppressor proteins such as p53, plO7, and Rb (retinoblastoma protein) have also been functionally linked to the SWI/SNF complex, as have proteins involved in DNA repair, including BRCAl and Fanconi's anemia protein.
  • loss of the BRM protein will strip away many of the mechanisms that are responsible for the control and fidelity of normal proliferation.
  • SWI/SNF complex In mammalian cells, numerous transcription factors, including Ets-2, ELICF, AP-I and Stat-3 require the SWI/SNF complex. Through these and other interactions, the SWI/SNF complex is important for the normal expression of a variety of genes. In yeast, the Swi/Snf complex controls the expression of approximately 5-7% of the yeast genome.
  • restoring BRM expression in accordance with the methods and compositions of the present invention has clinical applications.
  • SWI/SNF activity is required for the function of both RAR and PPAR ⁇ . Since agonists of RAR and PPAR ⁇ are clinically utilized as antitumor agents, restoring BRM could, in certain embodiments, increase the number of patients who could benefit from these drugs. Moreover, it has been shown that BRM expression is lost in a subset of both prostate and breast cancers.
  • BRM re-expression could be used to allow for the restoration of hormone sensitivity to breast and prostate cancer patients who have become refractory to anti-hormone therapy.
  • loss of BRM expression and SWI/SNF activity may herald more aggressive forms of cancers.
  • the proteins involved in DNA repair, such as p53, BRCAl and Fanconi's anemia, and in cell adhesion, such as integrins, CD44 and E-cadherin, are also linked to the SWI/SNF complex.
  • re-expression of BRM by the methods and compositions of the present invention could be used to thwart neoplastic development by restoring DNA repair mechanisms and reducing tumor metastatic potential. Furthermore, restoring BRM expression has antiproliferative effects. While not necessary to understand to practice the present invention this may be one mechanism by which HDAC inhibitors are inhibitory and have clinical efficacy.
  • Histone Deacetylases Nucleosomes, the primary scaffold of chromatin folding, are dynamic macromolecular structures, influencing chromatin solution conformations.
  • the nucleosome core is made up of histone proteins, H2A, HB, H3 and H4.
  • Histone acetylation causes nucleosomes and nucleosomal arrangements to behave with altered biophysical properties.
  • the balance between activities of histone acetyl transferases (HATs) and deacetylases (HDACs) determines the level of histone acetylation. Acetylated histones cause relaxation of chromatin and activation of gene transcription, whereas deacetylated chromatin generally is transcriptionally inactive.
  • HDACs Eleven different HDACs have been cloned from vertebrate organisms. The first three human HDACs identified were HDAC 1, HDAC 2 and HDAC 3 (termed class I human HDACs), and HDAC 8 has been added to this list. More recently class II human HDACs, HDAC 4, HDAC 5, HDAC 6, HDAC 7, HDAC 9, and HDAC 10 have been cloned and identified. Additionally, HDAC 11 has been identified but not yet classified as either class I or class II. All share homology in the catalytic region. HDACs 4, 5, 7, 9 and 10 however, have a unique amino-terminal extension not found in other HDACs. This amino-terminal region contains the MEF2-binding domain.
  • HDACs 4, 5 and 7 have been shown to be involved in the regulation of cardiac gene expression and in particular embodiments, repressing MEF2 transcriptional activity.
  • the exact mechanism in which class II HDACs repress MEF2 activity is not completely understood.
  • HDAC binding to MEF2 inhibits MEF2 transcriptional activity, either competitively or by destabilizing the native, transcriptionally active MEF2 conformation.
  • class II HDACs require dimerization with MEF2 to localize or position HDAC in a proximity to histones for deacetylation to proceed.
  • the present invention is not limited by the type of histone deacetylase inhibitor that is used with the methods and composition of the present invention.
  • a variety of inhibitors for histone deacetylases have been identified. The proposed uses range widely, but primarily focus on cancer therapy. Compounds which inhibit histone deacetylase (HDACs) have been shown to cause growth arrest, differentiation and/or apoptosis of many different types of tumor cell in vitro and in vivo.
  • HDACs histone deacetylase
  • HDAC inhibitors generally fall into four general classes: 1) short-chain fatty acids (e.g., 4-phenylbutyrate and valproic acid); hydroxamic acids (e.g., SAHA, Pyroxamide, trichostatin A (TSA), oxamflatin and CHAPs, such as, CHAPl and CHAP 31); 3) cyclic tetrapeptides (e.g., Trapoxin A and Apicidin); 4) benzamides (e.g., MS-275); and other compounds such as SCRIPTAID. Examples of such compounds can be found in U.S. Pat. No. 5,369,108; U.S. Pat. No. 5,700,811; and U.S. Pat. No.
  • HDACs can be inhibited a number of different ways such as by proteins, peptides, and nucleic acids (including antisense and RNAi molecules). Methods are widely known to those of skill in the art for the cloning, transfer and expression of genetic constructs, which include viral and non- viral vectors, and liposomes.
  • Viral vectors include adenovirus, adeno- associated virus, retrovirus, vaccina virus and herpesvirus.
  • RNAi type inhibitors are provided in Glaser et al., Biochem. and Biophys. Res. Comm., 310:529-36, 2003, herein incorporated by reference in its entirety).
  • Other HDAC inhibitors are small molecules. Perhaps the most widely known small molecule inhibitor of HDAC function is Trichostatin A, a hydroxamic acid. It has been shown to induce hyperacetylation and cause reversion of ras transformed cells to normal morphology and induces immunsuppression in a mouse model. It is commercially available from BIOMOL Research Labs, Lie, Plymouth Meeting, PA.
  • HDAC inhibitors that may find use in the present invention: U.S. Pat. 6,706,686; U.S. Pat. 6,541,661; U.S. Pat. 6,638,530; U.S. Pat. 6,541,661; U.S. Pat. Pub. 2004/0077698; EP1426054; U.S. Pat. Pub. 2003/0206946; U.S. Pat. 6,825,317; U.S. Pat. Pub. 2004/0229889; WO0215921; U.S. Pat. 5,993,845; U.S. Pat. Pub. 2004/0224991; WO04046094; U.S. Pat. Pub. 2003/0129724; U.S. Pat.
  • HDAC inhibitors examples include, but is not limited to, trichostatin A, trapoxin A, trapoxin B, HC-toxin, chlamydocin, Cly-2, WF-3161, Tan- 1746, apicidin, analogs of apicidin, benzamide, derivatives of benzamide, hydroxyamic acid derivatives, azelaic bishydroxyamic acid, butyric acid and salts thereof, actetate salts, suberoylanilide hydroxyamide acid, suberic bishydroxyamic acid, m-carboxy-cinnamic acid bishyrdoxyamic acid, oxamflatin, depudecin, tabucin, valproate, AN-9, CI-994, FR901228, and MS-27-275.
  • the agent can be a therapeutically effective oligonucleotide that inhibits expression or function of histone deacetylase, or a dominant negative fragment or variant of histone deacety
  • MethylGene Corp. such as Compound MGCD0103, and compounds LBH589 and LAQ824 from Novartis (see Qian et al., Clin. Cancer. Res., 2006, 12(2):634-42; and Remiszewski et al, J. Med. Chem., 2003, 46(21):4609-24), both of which are herein incorporated by reference.
  • Other preferred compounds are from Chroma therapeutics, such as Compound CHR-2504.
  • Table 1 provides additional HDAC inhibitors and the sensitivity known HDACs to these HDAC inhibitors.
  • the present invention provides methods for screening compounds, preferably
  • HDAC inhibitors to identify compounds that cause BRM expression.
  • the screening methods are not limited by the types of cells, but preferably employ cells that have reduced or absent BRM expression.
  • the cells employed not only have reduced BRM expression, but also have reduced levels of BRGl expression (i.e. reduced wild-type BRGl protein or mRNA expression levels).
  • the cells are contacted with a candidate compound (e.g. a HDAC inhibitor) and the expression of BRM mRNA and/or BRM protein is detected to determine if the compound causes an increase in such BRM expression.
  • a candidate compound e.g. a HDAC inhibitor
  • the host cells may already contain molecules that indicate the level of BRM mRNA expression or BRM protein expression such that no additional reagents need to be added to the cells.
  • the cells may be stably transfected with nucleic acid sequences for mRNA detection assays such as at least one of the following assays: the INVADER assay, a TAQMAN assay, a sequencing assay, a polymerase chain reaction assay, a hybridization assay, a hybridization assay employing a probe complementary to a mutation, a microarray assay, a bead array assay, a primer extension assay, an enzyme mismatch cleavage assay, a branched hybridization assay, a rolling circle replication assay, a NASBA assay, a molecular beacon assay, a cycling probe assay, a ligase chain reaction assay, and a sandwich hybridization assay.
  • one of these mRNA detection assays can be added to the cells after exposure to the candidate compound to determine if the compound caused an increase in BRM mRNA expression.
  • Responses of cells to treatment with the compounds can be detected by methods known in the art, including, but not limited to, fluorescence microscopy, confocal microscopy (e.g., FCS systems), flow cytometry, microfluidic devices, FLIPR systems (See, e.g., Schroeder and Neagle, J. Biomol. Screening 1:75 [1996]), and plate-reading systems.
  • the response e.g., increase in fluorescent intensity
  • the maximum response caused by a known agonist is defined as a 100% response.
  • the maximal response recorded after addition of an agonist to a sample containing a known or test antagonist is detectably lower than the 100% response.
  • the presence of BRM protein is detected in the cells after being contacted with a candidate compound.
  • Techniques for measuring such expression levels are known in the art.
  • One preferred technique is an ELISA assay that could employ antibodies directed to BRM to indicate the level of BRM expression after the cell is contacted with a candidate compound.
  • anti-BRM antibodies include, but are not limited to, the anti-BRM monoclonal antibody distributed by BD Biosciences (BD Biosciences, Franklin Lakes, NJ), and two anti-BRM polyclonal antibodies from Santa Cruz Biotechnology (Santa Cruz, CA).
  • test compounds can be obtained, for example, using any of the numerous approaches in combinatorial library methods known in the art, including biological libraries; peptoid libraries (libraries of molecules having the functionalities of peptides, but with a novel, non-peptide backbone, which are resistant to enzymatic degradation but which nevertheless remain bioactive; see, e.g., Zuckennann et al, J. Med. Chem. 37: 2678 [1994]); spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one- compound 1 library method; and synthetic library methods using affinity chromatography selection.
  • the biological library and peptoid library approaches are preferred for use with peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam (1997) Anticancer Drug Des. 12:145).
  • HDAC inhibitors are identified that are BRM expression- promoting histone deacetyalse inhibitors.
  • such inhibitors that only inhibit one of the known 11 HDACs, but still promote BRM expression, are identified.
  • various methods may be used. For example, RNAi may be used to selectively inhibit each of the 11 HDACs (e.g. one at a time) to determine which HDAC or HDACs can be inhibited and lead to BRM expression (e.g. lead to BRM expression in a cell deficient in BRM expression).
  • screening methods are employed to identify HDAC inhibitors that promote BRM expression, such that the BRM expressed is able to form part of a functioning SWI/SNF complex. For example, methods are employed that identify HDAC inhibitors that do not also induce the acetylation of BRM (as acetylation of BRM causes BRM to be inactivated).
  • CD44 and vimentin are detected as indicators of active BRM expression
  • Rb growth inhibition is detected. For example, to measure Rb growth inhibition, one could co- transfect MS-Rb, a constitutively active form of RB, in conjunction with a given HDAC inhibitor (e.g. a particular small molecule or siRNA). After 48 hours, transfected cells could be pulsed with BrdU for 24 hours and growth inhibition could be measured by immunostaining for BrdU incorporation.
  • the present invention provides therapeutic methods and compositions for treating a subject with a compound that promotes BRM expression in cancer cells in the patient that have reduced BRM expression.
  • the therapeutic compound is a HDAC inhibitor
  • the therapeutic compound is an HDAC inhibitor that specifically inhibits only one HDAC.
  • the HDAC inhibitor promotes expression of active BRM in cancer cells.
  • BRM peptides or nucleic acids sequences encoding BRM are administered to a patient.
  • the therapeutic compounds, peptides and nucleic acids of the present invention may be administered alone or in combination with at least one other agent, such as a stabilizing compound, and may be administered in any sterile, biocompatible pharmaceutical carrier, including, but not limited to, saline, buffered saline, dextrose, and water.
  • Peptides can be administered to the patient intravenously in a pharmaceutically acceptable carrier such as physiological saline.
  • Standard methods for intracellular delivery of peptides can be used (e.g., delivery via liposome). Such methods are well known to those of ordinary skill in the art.
  • the formulations of this invention are useful for parenteral administration, such as intravenous, subcutaneous, intramuscular, and intraperitoneal.
  • Therapeutic administration of a polypeptide intracellularly can also be accomplished using gene therapy methods. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and interaction with other drugs being concurrently administered.
  • these pharmaceutical compositions may be formulated and administered systemically or locally.
  • Techniques for formulation and administration may be found in the latest edition of "Remington's Pharmaceutical Sciences” (Mack Publishing Co, Easton Pa.). Suitable routes may, for example, include oral or transmucosal administration; as well as parenteral delivery, including intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, or intranasal administration.
  • compositions of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline.
  • physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline.
  • penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the pharmaceutical compositions of the present invention can be formulated using pharmaceutically acceptable carriers well known in the art in dosages suitable for oral administration.
  • Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral or nasal ingestion by a patient to be treated.
  • these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically.
  • the preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.
  • the pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known ⁇ e.g. , by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes).
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • compositions for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are carbohydrate or protein fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, etc; cellulose such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; and gums including arabic and tragacanth; and proteins such as gelatin and collagen.
  • disintegrating or solubilizing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings such as concentrated sugar solutions, which may also contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for product identification or to characterize the quantity of active compound, (i.e., dosage).
  • compositions that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients mixed with a filler or binders such as lactose or starches, lubricants such as talc or magnesium stearate, and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycol with or without stabilizers.
  • compositions comprising a compound of the invention formulated in a pharmaceutical acceptable carrier may be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • conditions indicated on the label may include treatment of condition related to apoptosis.
  • the pharmaceutical composition may be provided as a salt and can be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
  • the preferred preparation may be a lyophilized powder in 1 mM-50 mM histidine, 0.1%-2% sucrose, 2%-7% mannitol at a pH range of 4.5 to 5.5 that is combined with buffer prior to use.
  • the therapeutically effective dose can be estimated initially from cell culture assays. Then, preferably, dosage can be formulated in animal models (particularly murine models) to achieve a desirable circulating concentration range. With respect to HDAC inhibitors specifically, in certain embodiments, it is preferably administered at a sufficient dosage to attain a blood level of the inhibitor from about 0.01 M to about 100 M, more preferably from about 0.05 M to about 50 M, still more preferably from about 0.1 M to about 25 M, and still yet more preferably from about 0.5 M to about 25 M. For localized administration, much lower concentrations than this may be effective, and much higher concentrations may be tolerated. One of skill in the art will appreciate that the dosage of histone deacetylase inhibitor necessary to produce a therapeutic effect may vary considerably depending on the tissue, organ, or the particular animal or patient to be treated.
  • the therapeutic is a nucleic acid sequence encoding a HDAC inhibitor (e.g. siRNA, see, Glaser et al., Biochemical and Biophysical Res. Comm., 310:529-36, 2003, herein incorporated by reference) or a nucleic acid sequence encoding BRM.
  • the nucleic acid sequence is part of a vector such as an Adenovirus or Adeno-Associate virus such that the vector can express the nucleic acid sequence in the cells of a patient (e.g. cancer cells of a patient that are deficient for BRM expression).
  • the present invention provides methods and composition for treating viral infections, particularly in cancer patients. It is contemplated that many cancer patients actually die or get severely sick from cancer induced viral infections, rather than from their cancer, as their cancer leaves them exposed to such viral infections. Indeed, a large percent of cancer patients (e.g. 5% or more) may get sick or die from viral infections as a result of their cancer. While the cause of death may be officially noted as cancer, the true cause is actually viral infection that resulted from the cancer.
  • the present invention addresses this widespread problem by treating cancer patients to reduce their risk of cancer induced viral infection, or to help treat on-going viral infections that resulted from having cancer.
  • a cancer patient may have cancer cells that have reduced expression of BRM and/or interferon induced genes. Such reduced expression, it is contemplated, leaves the patient exposed to greatly increased risk of viral infection that may ultimately lead to severe sickness or death.
  • a patient is treated with compounds that increase the expression of at least one and preferably more interferon induced genes.
  • the present invention is not limited by the type of compound employed. Exemplary interferon induced genes that may be up-regulated to treat cancer induced viral infection are shown in Table 4.
  • the patient is treated with a histone deacetylase inhibitor in order to increase the expression of one or more interferon induced genes.
  • the patient is treated with BRM proteins or nucleic acid sequences that direct the expression of BRM proteins.
  • the present invention provides compositions and methods for detecting polymorphisms, such as SNPs and insertions, that provide information on whether SWI/SNF complexes will properly form or not in a given cell or population of cells, hi certain embodiments, polymorphisms in the BRM gene (including the promoter) are detected. In other embodiments, polymorphisms in the BRGl gene are detected. In some embodiments, nucleic acid detection assays are used to determine the presence or absence of polymorphisms in the BRM gene (including the promoter), such as position 741 insertions in the promoter.
  • nucleic acid detection assays are used to determine the presence or absence of polymorphisms in the BRGl gene, such as P311S; P316S; P319S, and P327S or other polymorphisms shown in Figure 1.
  • the present invention is not limited by the type of nucleic acid detection assay used to detect such polymorphisms. Detailed below are exemplary nucleic acid detection assays.
  • BRM and BRGl polymorphisms are detected using a direct sequencing technique.
  • nucleic acid samples are first isolated from a sample from a subject using any suitable method.
  • the region of interest is cloned into a suitable vector and amplified by growth in a host cell (e.g., a bacteria).
  • nucleic acid in the region of interest is amplified using PCR.
  • nucleic acid in the region of interest is sequenced using any suitable method, including but not limited to manual sequencing using radioactive marker nucleotides, or automated sequencing. The results of the sequencing are displayed using any suitable method. The sequence is examined and the presence or absence of BRM or BRGl polymorphisms are located.
  • BRM and BRGl polymorphisms are detected using a PCR-based assay, hi some embodiments, the PCR assay comprises the use of oligonucleotide primers that hybridize only to a given polymorphic sequence and primers that will not hybridize to the polymorphic sequence. Both sets of primers are used to amplify a sample of DNA. If only the polymorphic specific primers result in a PCR product, then the patient has the particular polymorphism.
  • BRM and BRGl polymorphisms are detected using a fragment length polymorphism assay.
  • a fragment length polymorphism assay a unique DNA banding pattern based on cleaving the DNA at a series of positions is generated using an enzyme (e.g., a restriction enzyme). Nucleic acid fragments from a sample containing a particular polymorphism will have a different banding pattern than those sequences not containing that particular polymorphism. 4.
  • BRM and BRGl polymorphisms are detected with a hybridization assay.
  • a hybridization assay the presence of absence of a particular polymorphism may be determined based on the ability of the nucleic acid from the sample to hybridize to a complementary nucleic acid molecule (e.g., an oligonucleotide probe).
  • a complementary nucleic acid molecule e.g., an oligonucleotide probe.
  • hybridization of a probe to the sequence of interest is detected directly by visualizing a bound probe (e.g., a Northern or Southern assay; See e.g., Ausabel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY [1991]).
  • a bound probe e.g., a Northern or Southern assay; See e.g., Ausabel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY [1991].
  • nucleic acid is isolated from a sample.
  • the DNA or RNA is then separated (e.g., on an agarose gel) and transferred to a membrane.
  • a labeled (e.g., by incorporating a radionucleotide) probe or probes specific for a BRM or BRGl polymorphism e.g. 7 base pair insertion at position 741 of the BRM promoter
  • Unbound probe is removed and the
  • BRM and BRGl related polymorphisms are detected using a DNA chip hybridization assay.
  • a DNA chip hybridization assay a series of oligonucleotide probes are affixed to a solid support. The oligonucleotide probes are designed to be unique to a given sequence.
  • the DNA sample of interest is contacted with the DNA "chip” and hybridization is detected.
  • the DNA chip assay is a GeneChip (Affymetrix, Santa Clara, CA; See e.g., U.S. Patent Nos.
  • Probe arrays are manufactured by Affymetrix's light directed chemical synthesis process, which combines solid phase chemical synthesis with photolithographic fabrication techniques employed in the semiconductor industry. Using a series of photolithographic masks to define chip exposure sites, followed by specific chemical synthesis steps, the process constructs high density arrays of oligonucleotides, with each probe in a predefined position in the array. Multiple probe arrays are synthesized simultaneously on a large glass wafer.
  • the wafers are then diced, and individual probe arrays are packaged in injection molded plastic cartridges, which protect them from the environment and serve as chambers for hybridization.
  • the nucleic acid to be analyzed is isolated, amplified by PCR, and labeled with a fluorescent reporter group.
  • the labeled DNA is then incubated with the array using a fluidics station.
  • the array is then inserted into the scanner, where patterns of hybridization are detected.
  • the hybridization data are collected as light emitted from the fluorescent reporter groups already incorporated into the target, which is bound to the probe array. Probes that perfectly match the target generally produce stronger signals than those that have mismatches. Since the sequence and position of each probe on the array are known, by complementarity, the identity of the target nucleic acid applied to the probe array can be determined.
  • a DNA microchip containing electronically captured probes (Nanogen, San Diego, CA) is utilized (See e.g., U.S. Patent Nos. 6,017,696; 6,068,818; and 6,051,380; each of which are herein incorporated by reference).
  • Nanogen's technology enables the active movement and concentration of charged molecules to and from designated test sites on its semiconductor microchip.
  • DNA capture probes unique to a given SNP or mutation are electronically placed at, or "addressed" to, specific sites on the microchip. Since DNA has a strong negative charge, it can be electronically moved to an area of positive charge.
  • an array technology based upon the segregation of fluids on a flat surface (chip) by differences in surface tension is utilized (See e.g., U.S. Patent Nos. 6,001,311; 5,985,551; and 5,474,796; each of which is herein incorporated by reference).
  • Protogene's technology is based on the fact that fluids can be segregated on a flat surface by differences in surface tension that have been imparted by chemical coatings. Once so segregated, oligonucleotide probes are synthesized directly on the chip by ink jet printing of reagents.
  • the array with its reaction sites defined by surface tension is mounted on a X/Y translation stage under a set of four piezoelectric nozzles, one for each of the four standard DNA bases.
  • the translation stage moves along each of the rows of the array and the appropriate reagent is delivered to each of the reaction site.
  • the A amidite is delivered only to the sites where amidite A is to be coupled during that synthesis step and so on.
  • Common reagents and washes are delivered by flooding the entire surface and then removing them by spinning.
  • DNA probes unique for positions BRM or BRGl polymorphisms are affixed to the chip using Protogene's technology. The chip is then contacted with the sample potentially containing nucleic acid sequences that may contain such polymorphisms. Following hybridization, unbound DNA is removed and hybridization is detected using any suitable method (e.g., by fluorescence de-quenching of an incorporated fluorescent group).
  • a "bead array” is used for the detection of BRM and BRGl polymorphisms (Illumina, San Diego, CA; See e.g., PCT Publications WO 99/67641 and WO 00/39587, each of which is herein incorporated by reference).
  • Illumina uses a BEAD ARRAY technology that combines fiber optic bundles and beads that self assemble into an array. Each fiber optic bundle contains thousands to millions of individual fibers depending on the diameter of the bundle.
  • the beads are coated with an oligonucleotide specific for particular BRM or BRGl polymorphisms. Batches of beads are combined to form a pool specific to the array.
  • the BEAD ARRAY is contacted with a prepared subject sample (e.g., DNA). Hybridization is detected using any suitable method.
  • hybridization is detected by enzymatic cleavage of specific structures (e.g., INVADER assay, Third Wave Technologies; See e.g., U.S. Patent Nos. 5,846,717; 5,985,557; 5,994,069; 6,001,567;
  • specific structures e.g., INVADER assay, Third Wave Technologies; See e.g., U.S. Patent Nos. 5,846,717; 5,985,557; 5,994,069; 6,001,567;
  • the INVADER assay detects specific DNA and RNA sequences by using structure specific enzymes to cleave a complex formed by the hybridization of overlapping oligonucleotide probes. Elevated temperature and an excess of one of the probes enable multiple probes to be cleaved for each target sequence present without temperature cycling. These cleaved probes then direct cleavage of a second labeled probe.
  • the secondary probe oligonucleotide can be 5' end labeled with a fluorescent dye that is quenched by a second dye or other quenching moiety.
  • the de-quenched dye-labeled product may be detected using a standard fluorescence plate reader, or an instrument configured to collect fluorescence data during the course of the reaction (i.e., a "real-time" fluorescence detector, such as an ABI 7700 Sequence Detection System, Applied Biosystems, Foster City, CA).
  • two oligonucleotides hybridize in tandem to the target nucleic acid to form an overlapping structure.
  • a structure-specific nuclease enzyme recognizes this overlapping structure and cleaves the primary probe, hi a secondary reaction, cleaved primary probe combines with a fluorescence-labeled secondary probe to create another overlapping structure that is cleaved by the enzyme.
  • the initial and secondary reactions can ran concurrently in the same vessel. Cleavage of the secondary probe is detected by using a fluorescence detector, as described above.
  • the signal of the test sample maybe compared to known positive and negative controls.
  • Additional detection assays that are produced and utilized using the systems and methods of the present invention include, but are not limited to, enzyme mismatch cleavage methods (e.g., Variagenics, U.S. Pat. Nos. 6,110,684, 5,958,692, 5,851,770, herein incorporated by reference in their entireties); polymerase chain reaction; branched hybridization methods (e.g., Chiron, U.S. Pat. Nos. 5,849,481, 5,710,264, 5,124,246, and 5,624,802, herein incorporated by reference in their entireties); rolling circle replication (e.g., U.S. Pat. Nos.
  • a MassARRAY system (Sequenom, San Diego, CA.) is used to detect BRM and BRGl related polymorphisms (See e.g., U.S. Patent Nos. 6,043,031; 5,777,324; and 5,605,798; each of which is herein incorporated by reference).
  • DNA is isolated from blood samples using standard procedures.
  • specific DNA regions containing the region of interest e.g, about 200 base pairs in length
  • the amplified fragments are then attached by one strand to a solid surface and the non immobilized strands are removed by standard denaturation and washing. The remaining immobilized single strand then serves as a template for automated enzymatic reactions that produce genotype specific diagnostic products.
  • Very small quantities of the enzymatic products are then transferred to a SpectroCHIP array for subsequent automated analysis with the SpectroREADER mass spectrometer.
  • Each spot is preloaded with light absorbing crystals that form a matrix with the dispensed diagnostic product.
  • the Mass ARRAY system uses MALDI TOF (Matrix Assisted Laser Desorption Ionization Time of Flight) mass spectrometry.
  • the matrix is hit with a pulse from a laser beam. Energy from the laser beam is transferred to the matrix and it is vaporized resulting in a small amount of the diagnostic product being expelled into a flight tube.
  • the diagnostic product As the diagnostic product is charged when an electrical field pulse is subsequently applied to the tube they are launched down the flight tube towards a detector.
  • the time between application of the electrical field pulse and collision of the diagnostic product with the detector is referred to as the time of flight.
  • This is a very precise measure of the product's molecular weight, as a molecule's mass correlates directly with time of flight with smaller molecules flying faster than larger molecules.
  • the entire assay is completed in less than one thousandth of a second, enabling samples to be analyzed in a total of 3-5 second including repetitive data collection.
  • the SpectroTYPER software then calculates, records, compares and reports the genotypes at the rate of three seconds per sample.
  • the present invention provides probes and primers specific for the BRM promoter (e.g. as shown in Figure 5).
  • the probes and primers are useful in detecting a polymorphism at position 741, and particularly the seven base pair insert TATTTTT (SEQ ID NO:42) shown in Figure 5.
  • Exemplary probes and primers that could be used with a nucleic acid detection assay such as those discussed above, include nucleic acids comprising, or consisting of, the following sequences: CTTTTCtatttttTATTTTT (SEQ ID NO:44); CCTTTTCtatttttTATTTTT (SEQ ID NO:45); CTTTTCtatttttTATTTTTT (SEQ ID NO:46); CCTTTTCtatttttTATTTTTT (SEQ ID NO:47); tatttttTATTTTTTATT (SEQ ID NO:48); tatttttTATTTTTTATTTT (SEQ ID NO:49); GCCCGCCTCCCTTTTCtattttt (SEQ ID NO:50); and CGCCTCCCTTTTCtattttt (SEQ ID NO:51).
  • the present invention provides PCR primers for amplifying the region surrounding the seven base pair insert shown in Figure 5.
  • PCR primers can be designed by generating at least one primer upstream of the seven base pair insert and at least one primer downstream of the seven base pair insert.
  • nested PCR primers are generated (e.g. two upstream primers and two downstream primers).
  • the present invention provides compositions comprising an isolated nucleic sequence that comprises SEQ ID NO: 52 (CCCTTTTCatttttT ATTTT TTATTTT).
  • Such nucleic acid sequence can be used, for example, as a positive control target in a nucleic acid detection assay designed to detect the seven base pair insert shown in Figure 5 or as a probe for detecting this seven base pair insert.
  • N normal
  • M molar
  • mM millimolar
  • ⁇ M micromolar
  • mol molecular weight
  • mmol millimoles
  • nmol nanomoles
  • pmol picomoles
  • g grams
  • mg milligrams
  • ⁇ g micrograms
  • ng nanograms
  • 1 or L liters
  • ml ml
  • This example describes sequencing BRGl and BRM sequences in cells lines deficient in BRGl and BRM protein expression. By western blotting, 10 cell lines were identified which lack BRGl and/or BRM expression. The characteristics of these cells lines are provided in Table 1.
  • BRGl and BRM mRNA transcript from each of these cell lines were sequenced.
  • a series of nested-PCR primer pairs that yield 5 overlapping PCR products spanning the coding region of each gene were employed. These primer pairs are shown in Table 2.
  • GRPSP APPAVPP AASPVMPP SEQ ID NO:41
  • SH3 Src homology 3
  • the H513 cell line had a similar splice variant, which deletes a 718 bp region extending from exon 4 to exon 6. In each of these cases, these variant transcripts disrupted the normal reading frame. As these cell lines lack any appreciable amount of the normal transcript, the changes likely abrogate the expression of this gene.
  • BRGl a variety of mutations were found that could account for the loss of expression in 7 out of the 10 cell lines, with only the Panc-1, C33a and Hl 573 lacking discernable abrogating mutations.
  • none of the ten cell lines demonstrated any significant alterations in BRM that could account for loss of expression. Specifically, nonsense mutations, insertions, deletions, or splicing variants were not detected.
  • This example describes the treatment of cells lines with undetectable BRGl and BRM protein expression with various HDAC inhibitors or 5-aza-deoxycytidine (5-azaCdR).
  • cell lines SW13, H522, H23 and A427 which have undetectable levels of BRGl/BRM proteins, were treated with DNA 5-aza-cytocytidine and sodium butyrate.
  • 5uM 5azaCytD was applied on three consecutive days, and then examined by semi- quantative RT-PCR the expression of pl6 in cell lines. Consistent with previous published reports, the silencing of pl6 in H23 and H441 cell lines were reversed with this treatment.
  • cell lines H522, SW13, A427, and H23 cell lines were treated with, trichostatin A, MS-275, or CI-994.
  • Treatment with 10 ⁇ M or 100 ⁇ M of MS-275 did not greatly affect BRM expression, but at a concentration of >1 niM, a modest induction of BRM was observed that was most robust in the A427cell line.
  • This lack of a strong induction effect, as compared to that of butyrate, is in part due to the increased toxicity of MS-275, which was most pronounced in the H23 and SW13 cell lines.
  • Treatment with either 600 riM of trichostatin or with 5uM of HDAC inhibitor CI-994 was also effective in inducing BRM expression in each of these cell lines.
  • the BRM promoter was cloned and its activity measured in BRGl/BRM positive and negative cell lines.
  • location of the BRM promoter was assessed by reviewing the location of available ESTs and capped cDNAs. This data showed that the BRM gene contains two first exons that are in tandem and upstream of exon2 where the translation start is located.
  • a screen for their expression of RT-PCR was performed. Using plasmids containing BRMlA or BRMlB cDNAs as standards, one was able to detect BRMlA mRNA but not BRMlB mRNA by RT-PCR.
  • both a 741bp and a 2.4kd DNA fragment was closed just upstream of exon IA into the pGL3 luficerase reporter vector. Transfecting these DNA fragment in both orientation in Calu3 and A549 yielded only robust luficerase activity when the promoters where in the correct orientation. Minimal luficerase activity was also noted with the control pGL3 in these cell lines.
  • the BRM promoter was sequenced in the 10 BRG1/BRM deficient cell lines. The several cell lines show a short insert which did appreciably altered luficerase activity when tested in Calu-6 or A549 BRM positive cell lines.
  • butyrate will promote histone acetylation by inhibiting the activity of a variety of HDACs , it is also know to promote the acetylation of varies other protein including p53 as well.
  • BRM epigenetic chromosome condensation of the BRM promoter
  • changes transcription factor activity mediate by histone acetylation
  • This Example describes an analysis of the temporal effects of HDAC inhibitors on BRM re-expression.
  • the time course at which BRM expression in SWl 3 cells was induced by continued exposure to butyrate was determined.
  • the upregulation of BRM expression was detected by western blot analysis at 12 hours and reached a plateau at 24 hours. Little change in BRM expression occurred with continued treatment for an additional 48 hours (Figure 3A).
  • This process was reversible, as BRM expression returned to pretreatment levels after removal of sodium butyrate.
  • SWl 3 cells were treated with butyrate for 72 hours, sodium butyrate was removed, and BRM levels were measured by western blotting from 0 to 6 days.
  • the BRM protein levels remained elevated for 72 hours and returned to near baseline levels at 96 hours ( Figure 3B).
  • the BRM mRNA level determined by quantative RT-PCR also remained elevated for 3 days, returning to baseline level by 4 days.
  • the Example describes a determination of which of the various common solid tumor types demonstrate the BRM deficiency.
  • six different high-density, tissue-specific microarrays were immunostained: lung, esophageal, ovarian, bladder, colon, and breast carcinomas, using a BRM-specific polyclonal antibody.
  • Anti-BRM antigen was prepared from the expression plasmid, pGEX-GST-BRM, containing a cDNA fragment of mouse BRM gene (encoding amino acid residues 50-214 in the corresponding human sequence) in pGEX-5X-2.
  • the GST-BRM fusion protein was expressed in E. coli BL21 and purified on a glutathione-Sepharose 4B column (Amersham, Piscataway, NJ) and GST-BRM fusion protein was used to produce rabbit polyclonal antibodies (Rockland, Rockland, MD).
  • BRM antisera was then passed over a GST-BRGl column to remove GST or BRGl reacting antibodies, and this negatively purified antisera was then further immunopurified by passing it over GST-BRM column.
  • BRM specificity and lack of BRGl cross reactivity of double affinity immunopurified antisera were confirmed by immunostaining paraffin embedded BRGl/BRM-deficient cell lines SWl 3 and H522 transfected with either BRGl or BRM.
  • the lung TMA was derived from surgery resection of pathological stage 1 and 2 cases at the University of Michigan from 1997-2001.
  • breast, colon, esophageal, bladder, and ovarian TMAs were constructed from University of Michigan surgical cases and were gifts from Drs. Kleer, Giordano, Beer, Shall and Cho, respectively.
  • TMA sections were deparaffinized with xylene and hydrated in a descending ethanol series to ddH2O. Before proceeding to antigen retrieval, sections were incubated 5 min in Ix PBS. Sections were immersed in 250 ml of 1OmM Tris-buffer, pH 10.0 in a covered plastic histology tank and placed in a microwaveable pressure cooker (Nordic Ware, Minneapolis, MN) containing 200 ml ddH2O. Sections were microwaved for 15 min at maximum power, then allowed to cool in the closed microwave for 10 min. After removal from the microwave, sections were slowly cooled in the sealed pressure cooker for 10 minutes under cold running water.
  • AU cases were reviewed by the pathologists in the study. Intensity of staining was defined as negative (no staining), weak (low staining), and positive (moderate and strong intensity) in over 80% of the tumor cells. All TMAs were reviewed blindly to clinical and pathological information.
  • BRM negative cases and 170 BRM positive nonsmall cell lung cancer cases the correlation between their differentiation states (poor, moderate and well) was examined by computing the independence test for each state of the two variables. The results showed a statistically insignificant result at the 5% level. From these data, it appears that the distribution of BRM-negative and -positive tumors is independent of differentiation state. Moreover, BRM expression was reduced in approximately 10% of esophageal cancers, but was retained in 31 Barrett's lesions examined, a precursor lesion for esophageal carcinoma, suggesting that BRM loss may not occur early in cancer development, but may be a hallmark of neoplastic transformation.
  • This Example describes methods used to test the role of BRM loss as it contributes to cancer progression.
  • An established experimental approach was employed that has previously been used to support the tumorigenic roles of such genes as Krev-1, p21, RASSFAl and Testin (see, e.g., Drusco et al, PNAS USA 102:10947-10951, 2005, and
  • mice are exposed to a known carcinogen and the differential effects on tumor occurrence are then studied.
  • BRM null mice were a gift from Moshe Yaniv, and their generation has been previously described (Miller et al., Cancer Lett, 198:139-144, 2003).
  • the BRM null mice are of 129/SV background and were crossed with 129/SV mice to BRM heterozygous mice.
  • Mice were treated at 8 weeks of age with intraperitoneal urethane lmg/kg and then monitored for tumor development in the lungs by sacrificing two mice from each group every 4 weeks. At 20 weeks, tumor development was observed in the control mice (BRM wild type mice).
  • mice in each group 10 per group were sacrificed and the effect of BRM expression on tumor development were compared by counting the number of visible surface tumors. It was found that a sequential increase in the number of tumors developing was a function of BRM allelic loss. Specifically, BRM wild-type mice had 2-3 tumors per mouse, whereas BRM heterozygous and BRM null mice had 12 and 25 tumors per mouse, respectively ( Figure 4B panel B). Similarly increased numbers were observed when cross-sections of the lungs of these animals were examined ( Figure 4C). However, a significant difference in tumor size or difference in histology type between these groups was not observed.
  • This Example describes methods used to analyze genes that are up-regulated upon reexpression of BRM.
  • microarray analysis was used to determine the identitity of genes that were up-regulated at least four-fold or more when BRM negative cell lines either SW13, A427 or NCI-H522 were transiently transfected with BRM (pCG-BRM vector) and a GFP expression vector and then were sorted by flow cytometry to selected for positively transfected subpopulation.
  • BRM negative cell lines either SW13, A427 or NCI-H522 were transiently transfected with BRM (pCG-BRM vector) and a GFP expression vector and then were sorted by flow cytometry to selected for positively transfected subpopulation.
  • BRM pCG-BRM vector
  • GFP expression vector a GFP expression vector
  • the genes are broken down into seven categories: i) differentiation genes; ii) tumor suppressor/oncogene/DNA repair genes; iii) cell adhesion genes; iv) extracellular matrix/structural genes; v) chemokine genes; vi) interferon-inducible genes; and vii) other genes.
  • LBH likely ortholog of mouse limb-bud and heart gene
  • CEACAMl carcinoembryonic antigen-related cell adhesion molecule 1 (biliary glycoprotein) CD44 CD44 antigen (homing function and Indian blood group system) CDHl cadherin 1, type 1, E-cadherin (epithelial) SPARCLl SPARC-like 1 (mast9, hevin)
  • PLAU plasminogen activator PLAU plasminogen activator, urokinase
  • CHI3L1 chitinase 3 -like 1 (cartilage glycoprotein-39)
  • SERPINB9 serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 9
  • P8 p8 protein (candidate of metastasis 1)
  • CHI3L1 chitinase 3-like 1 (cartilage glycoprotein-39)
  • TIMP3 tissue inhibitor of metalloproteinase 3 (Sorsby fundus dystrophy, pseudoinflammatory)
  • MATN2 matrilin 2 PLAT plasminogen activator tissue SVIL supervillin ITGA3 integrin, alpha 3 (antigen CD49C, alpha 3 subunit of VLA- 3 receptor)
  • IFIT3 interferon-induced protein with tetratricopeptide repeats 3
  • IFIT2 interferon-induced protein with tetratricopeptide repeats 2
  • IFIT3 interferon-induced protein with tetratricopeptide repeats 3
  • TGM2 transglutaminase 2 C polypeptide, protein-glutamine-gamma- glutamyltransferase
  • IFIT5 interferon-induced protein with tetratricopeptide repeats 5
  • G1P3 interferon, alpha-inducible protein (clone IFI-6- 16) ISG20 interferon stimulated gene 2OkDa
  • TIMP3 tissue inhibitor of metalloproteinase 3 (Sorsby fundus dystrophy, pseudoinflammatory)
  • ISGF3G interferon-stimulated transcription factor 3 gamma 48kDa
  • G1P2 interferon, alpha-inducible protein (clone IFI-15K)
  • TNFSFlO tumor necrosis factor (ligand) superfamily member 10 IGFBP6 insulin-like growth factor binding protein 6
  • SMARCA2 S WI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2
  • COLEC 12 collectin sub-family member 12
  • NCF2 neutrophil cytosolic factor 2 (65kDa, chronic granulomatous disease, autosomal 2)
  • HERC6 hect domain and RLD 6
  • ATP8B 1 ATPase Class I, type 8B, member 1
  • GPCR5A G protein-coupled receptor, family C, group 5, member A
  • ZC3HAV1 zinc finger CCCH type, antiviral 1
  • DDX58 DEAD (Asp-Glu-Ala-Asp) box polypeptide 58
  • PMAIPl phorbol- ⁇ -myristate-lS-acetate-induced protein 1
  • TNFSFlO tumor necrosis factor (ligand) superfamily member 10
  • PI3 protease inhibitor 3 skin-derived (SKALP)
  • PARP9 poly (ADP-ribose) polymerase family member 9
  • PARP 14 poly (ADP-ribose) polymerase family member 14 MX2 myxo virus (influenza virus) resistance 2 (mouse) nuclear antigen Sp 100 SP 100
  • FLJ22761 hypothetical protein FLJ22761
  • This Example describes the discovery of polymorphism in the human BRM promoter.
  • the presence of two polymorphisms within the BRM promoter have been discovered.
  • Each polymorphism is a 7 or 6 base pair insertion located at base pairs 741 and 1321 respectively.
  • the sequence of the 7 base pair insertion at position 741 was determined to be TATTTTT (SEQ E) NO:42), and the 6 base pair insertion at position 1321 was determined to be TTTTAA (SEQ DD NO:43).
  • the polymorphism at 741 strongly correlates with the loss of BRM expression while the 1321 does not.
  • tumors arising from individuals which are heterozygous at 741 lose the wild type allele and thus become functionally homozygous for the BRM 741 polymorphism. Therefore, the tumors are likely silencing BRM by losing the wild type allele and then by silencing the 741 allele via the aberrant recruitment of HDACs.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Toxicology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention a pour objet des procédés de criblage visant à identifier les inhibiteurs d’histone désacétylase (HDAC) favorisant l’expression des BRM, des procédés diagnostiques visant à déterminer l’applicabilité à un candidat sujet d’un traitement par un inhibiteur d’HDAC favorisant l’expression des BRM et des procédés thérapeutiques pour le traitement des cellules cancéreuses d’un patient par un inhibiteur d’HDAC favorisant l’expression des BRM. La présente invention a également pour objet des diagnostics de BRG1 et de BRM, des procédés pour renforcer la résistance d’un patient cancéreux aux infections virales et des procédés pour déterminer l’applicabilité à un candidat sujet d’un traitement par un composé glucocorticoïde ou à un composé rétinoïde.
PCT/US2006/007296 2005-03-01 2006-03-01 Inhibiteurs d’hdac favorisant l’expression des brm et diagnostics associes aux brm WO2006094068A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US65760305P 2005-03-01 2005-03-01
US60/657,603 2005-03-01

Publications (2)

Publication Number Publication Date
WO2006094068A2 true WO2006094068A2 (fr) 2006-09-08
WO2006094068A3 WO2006094068A3 (fr) 2009-04-16

Family

ID=36941784

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/007296 WO2006094068A2 (fr) 2005-03-01 2006-03-01 Inhibiteurs d’hdac favorisant l’expression des brm et diagnostics associes aux brm

Country Status (2)

Country Link
US (1) US7604939B2 (fr)
WO (1) WO2006094068A2 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011132175A2 (fr) 2010-04-22 2011-10-27 British Columbia Cancer Agency Branch Nouveaux marqueurs biologiques et cibles pour le carcinome ovarien
WO2012135588A2 (fr) * 2011-04-01 2012-10-04 Sloan Kettering Institute For Cancer Research Procédés de traitement d'un cancer séreux
US20140051635A1 (en) * 2011-02-02 2014-02-20 University Of South Alabama Combination therapy for treatment of cancer
US9308276B2 (en) 2011-05-17 2016-04-12 University Of South Alabama Combination therapy for treatment of cancer

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20090098920A (ko) * 2007-01-10 2009-09-17 노파르티스 아게 데아세틸라제 억제제의 제제
CL2008002786A1 (es) * 2007-09-20 2009-05-15 Novartis Ag Torta farmceuticamente aceptable, formada por liofilizacion, que comprende: n-hidroxi-3-[4-[[[2-(2-metil-1h-indol-3-il]-etil]-amino]-metil]-fenil]-2e-2-propenamida o una sal, un regulador de ph seleciondo de lactato o acidop lactico, fodfato o acido fosforico o una combinacion y un agente de volumen; proceso de elaboracion.
WO2016164482A1 (fr) * 2015-04-06 2016-10-13 The University Of North Carolina At Chapel Hill Méthodes et compositions pour le traitement de l'insuffisance cardiaque
CN113502300B (zh) * 2021-07-21 2024-02-06 徐州医科大学 一种含有hdac10基因启动子序列和报告基因的重组质粒、制备方法及应用

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5175191A (en) 1988-11-14 1992-12-29 Sloan-Kettering Institute For Cancer Research Potent inducers of terminal differentiation and methods of use thereof
US5055608A (en) 1988-11-14 1991-10-08 Sloan-Kettering Institute For Cancer Research Novel potent inducers of thermal differentiation and method of use thereof
US5700811A (en) 1991-10-04 1997-12-23 Sloan-Kettering Institute For Cancer Research Potent inducers of terminal differentiation and method of use thereof
US5369108A (en) 1991-10-04 1994-11-29 Sloan-Kettering Institute For Cancer Research Potent inducers of terminal differentiation and methods of use thereof
US5922837A (en) 1995-09-20 1999-07-13 Merck & Co., Inc. Antiprotozoal cyclic tetrapeptides
EP0827742A1 (fr) 1996-09-04 1998-03-11 Vrije Universiteit Brussel Utilisation des inhibiteurs de l'histone déacétylase pour le traitement de la fibrose ou de la cirrhose
JP4269041B2 (ja) 1999-03-02 2009-05-27 国立大学法人九州工業大学 新規な環状テトラペプチド誘導体とその医薬用途
JP2001081031A (ja) 1999-08-30 2001-03-27 Schering Ag 溶解性および経口吸収性を改善したベンズアミド誘導体含有製剤
EP1231919B1 (fr) 1999-09-08 2015-09-30 Sloan-Kettering Institute For Cancer Research Derivés de 1-amino-1-(hétero)arylaminocarbonyl-6-hydroxyaminocarbonylhexan utiles dans le traitement de tumeurs
KR20020070285A (ko) 1999-11-23 2002-09-05 메틸진, 인크. 히스톤 디아세틸라제의 억제제
US20030129724A1 (en) 2000-03-03 2003-07-10 Grozinger Christina M. Class II human histone deacetylases, and uses related thereto
WO2002015921A2 (fr) 2000-08-18 2002-02-28 The Governement Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Methodes de traitement du lymphome cutane a cellules t et du lymphome peripherique a cellules t (non specifie) par l'administration d'un inhibiteur de l'histone deacetylase
US20020103192A1 (en) 2000-10-26 2002-08-01 Curtin Michael L. Inhibitors of histone deacetylase
AR035417A1 (es) 2001-01-27 2004-05-26 Hoffmann La Roche Derivados triciclicos de lactama y sultama, procesos para su elaboracion, medicamentos que los contienen, y el uso de dichos compuestos en la preparacion de medicamentos
US20020177594A1 (en) 2001-03-14 2002-11-28 Curtin Michael L. Inhibitors of histone deacetylase
US6784173B2 (en) 2001-06-15 2004-08-31 Hoffmann-La Roche Inc. Aromatic dicarboxylic acid derivatives
US8470783B2 (en) 2001-08-21 2013-06-25 Astellas Pharma Inc. Medicinal use of histone deacetylase inhibitor and method of evaluating antitumor effect thereof
US6706686B2 (en) 2001-09-27 2004-03-16 The Regents Of The University Of Colorado Inhibition of histone deacetylase as a treatment for cardiac hypertrophy
US20040214860A1 (en) 2001-11-09 2004-10-28 Charous B. Lauren Method for treating inflammatory bowel disease
US20040132825A1 (en) 2002-03-04 2004-07-08 Bacopoulos Nicholas G. Methods of treating cancer with HDAC inhibitors
US20030206946A1 (en) 2002-04-26 2003-11-06 Yih-Lin Chung Methods for therapy of connective tissue disease
GB0226855D0 (en) 2002-11-18 2002-12-24 Queen Mary & Westfield College Histone deacetylase inhibitors
TW200418806A (en) 2003-01-13 2004-10-01 Fujisawa Pharmaceutical Co HDAC inhibitor
US7244751B2 (en) 2003-02-14 2007-07-17 Shenzhen Chipscreen Biosciences Ltd. Histone deacetylase inhibitors of novel benzamide derivatives with potent differentiation and anti-proliferation activity
US20060235012A1 (en) 2003-06-16 2006-10-19 Chroma Therapeutics Limited Carboline and betacarboline derivatives for use as hdac enzyme inhibitors
US7842835B2 (en) 2003-07-07 2010-11-30 Georgetown University Histone deacetylase inhibitors and methods of use thereof

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BOURACHOT ET AL.: 'Growth inhibition by the mammalian SWI-SNF subunit Brm is regulated by acetylation.' THE EMBO JOURNAL. vol. 22, no. 24, 2003, pages 6505 - 6515 *
MIZUTANI ET AL.: 'Maintenance of integrated proviral gene expression requires Brm, a catalytic subunit of SWI/SNF complex.' J OF BIOLOGICAL CHEMISTRY vol. 277, no. 18, May 2002, pages 15859 - 15864 *
REISMAN ET AL.: 'Concomitant down-regulation of BRM and BRG1 in human tumor cell lines: differential effects on RB-mediated growth arrest vs. CD44 expression.' ONCOGENE vol. 21, 2002, pages 1196 - 1207 *
WONG ET AL.: 'BRG I, a component of the SWI-SNF complex, is mutated in multiple human tumor cell lines.' CANCER RESEARCH. NOVEMBER vol. 60, 2000, pages 6171 - 6177 *
YAMAMICHI-NISHINA ET AL.: 'SWI3 cells can transition between two distinct subtypes by switching expression of BRG1 and Brm genes at the post-transcription level.' J OF BIOLOGICAL CHEMISTRY vol. 278, no. 9, February 2003, pages 7422 - 7430 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011132175A2 (fr) 2010-04-22 2011-10-27 British Columbia Cancer Agency Branch Nouveaux marqueurs biologiques et cibles pour le carcinome ovarien
EP2561351A2 (fr) * 2010-04-22 2013-02-27 British Columbia Cancer Agency Branch Nouveaux marqueurs biologiques et cibles pour le carcinome ovarien
CN103026227A (zh) * 2010-04-22 2013-04-03 不列颠哥伦比亚省癌症分社 新的卵巢癌生物标志物和靶
EP2561351A4 (fr) * 2010-04-22 2013-10-16 British Columbia Cancer Agency Nouveaux marqueurs biologiques et cibles pour le carcinome ovarien
US20140051635A1 (en) * 2011-02-02 2014-02-20 University Of South Alabama Combination therapy for treatment of cancer
WO2012135588A2 (fr) * 2011-04-01 2012-10-04 Sloan Kettering Institute For Cancer Research Procédés de traitement d'un cancer séreux
WO2012135588A3 (fr) * 2011-04-01 2012-12-27 Sloan Kettering Institute For Cancer Research Procédés de traitement d'un cancer séreux
US9308276B2 (en) 2011-05-17 2016-04-12 University Of South Alabama Combination therapy for treatment of cancer
US9827328B2 (en) 2011-05-17 2017-11-28 University Of South Alabama Combination therapy for treatment of cancer

Also Published As

Publication number Publication date
WO2006094068A3 (fr) 2009-04-16
US7604939B2 (en) 2009-10-20
US20060292594A1 (en) 2006-12-28

Similar Documents

Publication Publication Date Title
US20120122718A1 (en) BRM Expression and Related Diagnostics
US7604939B2 (en) Methods of identifying active BRM expression-promoting HDAC inhibitors
JP5649263B2 (ja) 心臓血管障害および薬物応答に関連した遺伝子多型、それらの検出方法および用途
JP5918216B2 (ja) スタチンの応答および心血管疾患に関連する遺伝子多型、その検出方法ならびに使用
EP2944961A1 (fr) Marqueurs pour le pronostic et la therapie du cancer et procedes d'utilisation
CA2726691C (fr) Utilisation de l'oncogene nrf2 aux fins d'un pronostic de cancer
US11579140B2 (en) Methods related to bronchial premalignant lesion severity and progression
US20060134664A1 (en) Identification of dysregulated genes in patients with neurological diseases
US20170275705A1 (en) Biomarkers useful for determining response to pd-1 blockade therapy
KR20080080525A (ko) 유전자 전사에 대한 fgfr3의 억제제의 효과
US20100143916A1 (en) Osteoarthritis biomarkers and uses thereof
WO2011133609A2 (fr) Méthodes et nécessaires de prédiction de résultat thérapeutique associée à l'utilisation d'inhibiteurs de la btk
JP2008514209A (ja) 癌マーカー
Liang et al. Novel missense mutations in PAX9 causing oligodontia
JP2008539728A (ja) 膀胱癌バイオマーカーおよびその使用
EP2711433B1 (fr) Méthode de prédiction de l'efficacité d'un inhibiteur de l'angiogenèse
US20070275925A1 (en) Asthma diagnosis and therapy
WO2009089102A2 (fr) Identification et caractérisation de signatures génétiques associées à la grossesse et leur utilisation pour le diagnostic et le traitement du cancer du sein
US20100087328A1 (en) Brm expression and related diagnostics
WO1999047692A2 (fr) Procedes de detection de troubles neurologiques
US20060172295A1 (en) Genes associated with schizophrenia adhd and bipolar disorders
WO2010014582A2 (fr) Expression des mrb et diagnostic associé
US9678060B2 (en) Lunasin-induced regulation of disease-related gene expression
US20070148666A1 (en) Diagnosis and Treatment of Brain Cancer
US20220291220A1 (en) Methods and compositions for detection and treatment of lung cancer

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 06736589

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 06736589

Country of ref document: EP

Kind code of ref document: A2